Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Oct 17.
Published in final edited form as: Curr Top Med Chem. 2015;15(21):2211–2221. doi: 10.2174/1568026615666150610142234

Nicotinamidephosphoribosyltransferase and cerebral ischemia

Xinzhi Chen 1, Shangfeng Zhao 1,2, Yang Song 1, Yejie Shi 1, Rehana Leak 1,3, Guodong Cao 1,4
PMCID: PMC5644507  NIHMSID: NIHMS875869  PMID: 26059356

Abstract

Recombinant tissue plasminogen activator is the only drug approved for the clinical treatment of acute ischemic stroke. Thus, there is an urgent unmet need for novel stroke treatments. Endogenous defense mechanisms against stroke may hold the key to new therapies for stroke. A large number of studies suggest that nicotinamidephosphoribosyl-transferase (NAMPT) is an attractive candidate to improve post-stroke recovery. As a multifunctional protein, NAMPT plays important roles in immunity, metabolism, aging, inflammation, and stress responses. NAMPT exists in both the intracellular and extracellular space. As a rate-limiting enzyme, intracellular form (iNAMPT) catalyzes the first step in the biosynthesis of nicotinamide adenine dinucleotide (NAD) from nicotinamide. iNAMPT closely regulates energy metabolism, enhancing the proliferation of endothelial cells, inhibiting apoptosis, regulating vascular tone, and stimulating autophagy in disease conditions such as stroke. Extracellular NAMPT (eNAMPT) is also known as visfatin (visceral fat–derived adipokine) and has pleotropic effects. It is widely believed that the diverse biological functions of eNAMPT are attributed to its NAMPT enzymatic activity. However, the effects of eNAMPT on ischemic injury are still controversial. Some authors have argued that eNAMPT exacerbates ischemic neuronal injury non-enzymatically by triggering the release of TNF-α from glial cells. In addition, NAMPT also participates in several pathophysiological processes such as hypertension, atherosclerosis, and ischemic heart disease. Thus, it remains unclear under what conditions NAMPT is beneficial or destructive. Recent work using in vitro and in vivo genetic/pharmacologic manipulations, including our own studies, has improved our understanding of NAMPT. This review focuses on the multifaceted and complex roles of NAMPT under normal and ischemic conditions.

Keywords: adipokine, brain ischemia, intracerebroventricular, mouse, neuroprotection, reperfusion injury, stroke, vascular biology

1. Introduction

Stroke is the second most common cause of death worldwide and the leading cause of long-term adult disability in the United States. Thus far, the only drug that has been used successfully to treat acute stroke in the clinic is the clot-dissolving drug tissue plasminogen activator (tPA). However, this medication is restricted in use because of the limited temporal window (within three hours of stroke symptom onset) during which it can be safely applied [1]. To date, numerous cytoprotective drugs have reached phase 3 efficacy trials in acute stroke patients. Unfortunately, throughout the clinical literature, the phrase “failure to demonstrate efficacy” prevails as a common thread, despite mostly encouraging results in preclinical studies (Labiche LA 2004 46). It is therefore imperative to develop new neuroprotective and neurorestorative strategies for ischemic stroke. Endogenous defense mechanisms against stroke injury may hold the key to the development of novel therapies for stroke. A large number of studies have suggested that nicotinamidephosphoribosyl-transferase (NAMPT) is an attractive candidate to improve post-stroke recovery. NAMPT is the rate-limiting enzyme in the salvage pathway of nicotinamide adenine dinucleotide (NAD) biosynthesis. Depletion of NAD due to energy depletion and PARP-1 consumption is a critical step in the induction of cell death after ischemia [24]. Thus, as the key regulator of NAD biosynthesis, NAMPT has been postulated to exert a significant impact on cellular function and survival under ischemic conditions. Besides regulating energy metabolism, recent studies have confirmed that NAMPT has multiple functions: enhancing the proliferation of endothelial cells, inhibiting apoptosis, regulating vascular tone, and stimulating autophagy. Lim et al. have shown that the administration of NAMPT recombinant protein at the time of reperfusion reduces infarct size by approximately 20% in a mouse model of cardiac ischemia [5]. Genetic deletion or pharmacological inhibition of NAMPT worsens ischemic brain injury [6] and lentivirus-mediated NAMPT overexpression protects against ischemic brain injury [7]. The precise neuroprotective mechanisms underlying the efficacy of NAMPT in cerebral ischemia remain unknown, although previous studies have suggested that NAMPT may mediate neuroprotection by providing extra energy and activating NAD-dependent survival signaling pathways. These studies provide rationale for investigating NAMPT in ischemic stroke. However, there is little agreement on the effects of NAMPT on ischemic injury. Some reports have argued that NAMPT exacerbates ischemic neuronal injury non-enzymatically by triggering the release of tumor necrosis factor alpha (TNF-α) from glial cells [8]. In addition, NAMPT also participates in several pathophysiological processes such as hypertension, atherosclerosis, and ischemic heart disease. Thus, the conditions under which NAMPT is beneficial remain unclear. Recent studies using in vitro and in vivo genetic/pharmacologic manipulations, including our own studies, have shed some light on the answer to this question. This review focuses on 1) basic characteristics of NAMPT that have the potential to be modified in pathological conditions, 2) various complex functions of NAMPT that tie in to ischemic conditions, and 3) the underlying mechanisms.

2. Basic characteristics of NAMPT

The human NAMPT (hNAMPT) gene (Gene ID: 10135) has 37,643 bp and is located on chromosome 7q22.3 according to Pubmed (2014). The structural NAMPT gene is composed of 11 exons and 10 introns. The regulatory gene at the 5′-flanking region has a 1.4 kb region that is GC rich (60% GC) following a 1.6 kb region with more AT bases (60% AT). The first 500 bp upstream of the major transcription initiation site (TIS) lacks canonical TATA and CAAT boxes but has higher G+C content (72% GC). Several CAAT boxes and TATA-like and initiator sequences are found about 2 kb upstream from the TIS, which may act as the promoter region. The binding sites for transcription factors such as nuclear factor 1 (NF1) and selective promoter factor 1 (Sp1) activator protein (AP-1/AP-2) and hepatocyte nuclear factors (HNFs) have been identified. The binding sites for the glucocorticoid receptor, corticotropin releasing factor, cAMP response element binding protein, and the nuclear factors (such as NF-IL6, NF-κB) are found in the promoter region. These multiple regulatory elements hint at diverse means of transcriptional modulation [9].

By December 24, 2014, 1480 SNPs in the hNAMPT gene have been reported. The functional consequences of most of these SNPs are currently unknown. In general, gene function may be affected when SNPs occur within a regulatory region near a gene [10]. Ye et al. discovered a 7.7-fold higher risk of acute respiratory distress syndrome (ARDS) with two SNPs (T-1001G and C-1543T) in the hNAMPT gene promoter, and TT represents a protective haplotype against ARDS [10]. Bajwa et al. found that the NAMPT T-1001G variant allele and related haplotype are associated with increased odds of developing ARDS and increased intensive care unit mortality among at-risk patients, whereas the C-1543T variant allele and related haplotype are associated with decreased odds of ARDS among patients with septic shock and better outcomes among patients with ARDS [11]. The G-948T gene polymorphism was associated with increased high-density lipoprotein (HDL) cholesterol in obese subjects [12]. The −1535T/T genotype was associated with lower serum triglyceride levels and higher HDL-cholesterol levels in non-diabetic Japanese subjects [13]. rs9770242 and rs1319501 were in perfect linkage disequilibrium, whereas fasting insulin and glucose levels and rs7789066 were significantly associated with the apolipoprotein B component of very low density lipoprotein (VLDL) from the Quebec Family Study [14]. rs9770242, −948G-T, and rs4730153 were associated with the ratio of visceral/subcutaneous visfatin mRNA expression [15]. In addition, −948G-T was associated with 2-h plasma glucose and fasting insulin concentrations in nondiabetic Caucasian subjects [15]. These studies reveal that the NAMPT gene may play an important role in metabolic function.

For the hNAMPT gene, transcription produces 19 different mRNAs, 14 alternatively spliced variants and 5 unspliced forms. There are 5 probable alternative promoters, 6 non-overlapping alternative last exons and 13 alternative polyadenylation sites. The mRNAs appear to differ by truncation of the 5′ end, truncation of the 3′ end, presence or absence of 2 cassette exons, overlapping exons with different boundaries, alternative splicing or retention of 4 introns. The physiological relevance of these NAMPT variants are currently unknown, and assumed to be even more complicated under pathological conditions.

NAMPT mRNA is ubiquitously distributed in all tissues including heart, brain, placenta, lungs, liver, skeletal muscle, kidney blood vessels, and pancreases. Liver and muscle tissue show the highest mRNA levels [9,1618].

NAMPT protein is highly conserved from invertebrates to mammals, including humans. The canine NAMPT protein sequence is 96% identical to the human sequence and 94% identical to murine and rat counterparts [19]. The mammalian gene is functionally active in prokaryotic hosts [20]. The hNAMPT protein sequence (NP_005737.1) consists of 491 amino acids with a predict molecular weight of 52 kDa. It contains 63 negatively charged residues (Asp + Glu) and 61 positively charged residues (Arg + Lys). hNAMPT is considered to be a stable protein, since the estimated half-life is 30 hours based on the methionine at the N-terminal of the sequence.

Several groups have resolved the three dimensional structure of mammalian NAMPT protein [21]. The structure of the NAMPT monomer has three domains containing total 22 β-strands and 15 α-helices. Homodimerized NAMPT catalyzes the conversion of nicotinamide (NAM) and phosphoribosyl-pyrophosphates to form nicotinoamide mononucleotide, a key step in the mammalian NAD synthetic salvage pathway. The 3D structure analysis of human and murine NAMPT by Khan et al. [22] showed that NAMPT is functionally similar to quinolinic acid and nicotinic acid phosphoribosyltransferases, enzymes in two other NAD biosynthesis pathways, despite having la arger size and highly divergent amino acid sequences.

NAMPT catalyzes ATP hydrolysis and become autophosphorylated at a histidine residue (His247) that lies at the center of a conserved cluster of active site residues. Autophosphorylation can increase its enzymatic activity and lower the Km for the substrates [23]. Wang et al reported that NAMPT is in the dimeric class of type II phosphoribosyltransferases with a unique hydrogen bond between Asp219 and the amide of nicotinamide. Mutations of the residue H247 significantly affect NAMPT enzymatic activity [24]. The structural and mutagenesis studies by Khan et al. [16] demonstrate that Asp219 is important in defining the substrate specificity of NAMPT. The structural differences between these enzymes indicate variations in substrate specificity and inhibitor sensitivity. Furthermore, although the 5′-flanking region of the gene of NAMPT lacks the classical secretion signal sequences [9], it can be secreted via an endoplasmic reticulum–Golgi or microvesicles independent pathway [25,26].

3. Regulation of NAMPT

The basic characteristics and regulation of NAMPT have been revealed by a plethora of studies using small molecule inhibitors, siRNA, miRNA, antisense oligonucleotide, gene overexpression, gene knock out, and antibodies. The current chemical inhibitors of NAMPT are analogues of FK866 (APO866, (E)-N-[4-(1-benzoylpiperidin-4-yl) butyl] acrylamide-3-(pyridin-3-yl) and CHS 828 (N-(6-chlorophenoxyhexyl)-Nt-cyano-Nu-4-pyridylguanidine).

The use of FK866 as a NAMPT inhibitor was first reported by Hasmann and Schemainda [27]. FK866 induced apoptosis in HepG2 human liver carcinoma cells by a highly specific and potent inhibition of NAMPT with an IC50 of approximately 1 nM [27]. FK866 did not directly inhibit mitochondrial respiratory activity, but caused gradual NAD+ depletion by inhibiting NAMPT, which eventually triggers apoptosis. Nicotinic acid and NAM were both found mitigate the cellular effects of FK866. The mechanism underlying FK866 inhibition of NAMPT has been well characterized. The crystal structure of NAMPT, free and in complex with FK866, has been identified by Wang et al [24] and Kim et al [28]. FK866 binds at the nicotinamide-binding site of NAMPT (a tunnel at the interface of the NAMPT dimer), thus functioning as a competitive inhibitor of NAMPT enzymatic activity [22,24,28]. Mutations in this binding site can abolish the inhibition by FK866. FK866 binds to the active site of NAMPT with higher affinity than either the substrate or the product. The benzoylpiperidin group of FK866 plays a key role in binding to NAMPT in hydrophobic interactions [28]. As NAMPT acts as a key enzyme of NAD synthesis, FK866 displays strong anticancer activity in hematologic malignancies both in vitro and in vivo [29,30].

CHS 828, a cyanoguanidine compound, is a structurally unique NAMPT inhibitor currently in phase II clinical trials for cancer patients [3134]. CHS 828 inhibits NAD synthesis in a similar way as FK866. Increasing cellular levels of NAD can completely block the cellular effects of CHS 828, suggesting a competitive mode of action. Furthermore, the crystal structure and in vitro biochemistry shows that FK866 and CHS 828 share a common binding site at the active site of NAMPT [28,35,36].

As mentioned in Section 2 (Basic characteristics of NAMPT), NAMPT can be upregulated in several different ways. The activation of NAMPT by H247-phosphorylation causes stabilization of the enzyme-phosphoribosylpyrophosphate complex, permitting efficient capture of nicotinamide. Small changes in distances in the reaction coordinates also have large effects on catalysis, and may account for the activation of NAMPT when phosphorylated at H247. The activation of NAMPT by ATP hydrolysis also suggests that physiological enzymatic activation is a consequence of the formation of phospho-His-247 (Burgos ES 2008 11086) [24]. Recent studies have identified NAMPT as the intracellular target of P7C3, a proneurogenic compound containing an aminopropyl carbazole. P7C3 enhance the enzymatic activity of purified NAMPT by increasing NAD levels through NAMPT-mediated salvage pathways. These studies leave unanswered how exactly P7C3 works, as the native NAMPT enzyme is regulated by posttranslational modifications, intracellular localization, and a wide spectrum of other intracellular variables (Wang g 2014 1324).

4. Physiological functions of NAMPT

Accumulating evidence indicates that NAMPT has at least three major functions–as a nicotinamidephosphoribosyltransferase, a growth factor, and a cytokine.

4.1 NAMPT functions as a nicotinamidephosphoribosyltransferase

Most of the available evidence indicates that biological actions mediated by NAMPT are mainly due to enzymatic function [3740]. NAD is an essential coenzyme in metabolic pathways, and is regarded as a universal energy- and signal-carrying molecule in the control of fundamental cellular processes, including signal transduction [41]. DNA repair [42], calcium homeostasis [43], gene regulation [44], longevity [45], genomic integrity [46] and apoptosis [4750]. Therefore, NAMPT plays an important role in a variety of physiological processes through the synthesis of NAD, such as metabolism, inflammation, and responses to stress [51].

Pre-B-cell colony-enhancing factor (PBEF) could be a NAMPT, as initially suggested when Martin et al. discovered that the sequence of the nadV gene is homologous to that of hNAMPT. The product encoded by this gene allows A. pleuropneumoniae to utilize nicotinamide mononucleotide as a precursor for NAD biosynthesis [52]. Rongvaux et al. have verified that PBEF is indeed a NAMPT. They demonstrated that the murine protein catalyzes the condensation of NAM with 5-phosphoribosyl-1-pyrophosphate to yield nicotinamide mononucleotide, an intermediate in the biosynthesis of NAD, similar to its microbial counterpart. The role of PBEF as a NAMPT was further confirmed by experiments showing that the mouse gene confers an NAMPT-defective bacterial strain the ability to grow in the absence of NAD [20]. The extracellular form of NAMPT (eNAMPT) exhibits robust NAD biosynthetic activity and regulates insulin secretion in beta cells during glucose homeostasis.

NAMPT positively regulates the activity of silent mating type information regulation 2 homolog 1 (SIRT1), a putative longevity protein [53,54]. Study by Revollo et al. [53] demonstrated that NAMPT catalyzes a rate-limiting step in a salvage pathway of the mammalian NAD biosynthesis. Van der Veer et al. demonstrated that the enhanced NAMPT activity of PBEF lengthens the cellular lifespan of human primary smooth muscle cells, human clonal smooth muscle cells, and fibroblasts derived from a patient with Hutchinson-Gilford progeria syndrome [54]. Introducing the NAMPT gene into aging human smooth muscle cells (SMCs) delays senescence and substantially lengthens cell lifespan, and enhances resistance to oxidative stress. NAMPT-mediated smooth muscle cell (SMC) lifespan extension is associated with increased activity of the NAD+-dependent longevity enzyme SIRT1. NAMPT overexpression also reduced the fraction of p53 that was acetylated on lysine 382 (a target of SIRT1), suppressed an age-related increase in p53 expression, and increased the rate of p53 degradation. Thus, NAMPT may also be a longevity protein that can add stress-resistance and extend the lifespan of human SMCs by optimizing SIRT1-mediated p53 degradation. NAMPT also has anti-apoptotic effects in many cell types [37,39,5456]. Recently, Benigi et al. reported that the longevity phenotype in angiotension II type 1 receptor knockout mice was associated with an increased number of mitochondria and upregulation of the prosurvival genes NAMPT and sirtuin 3 (Sirt3) in mouse proximal tubular epithelial cells [57]. The authors postulate that disruption of angiotension II type 1 receptor promotes longevity in mice, possibly by the attenuation of oxidative stress and overexpression of prosurvival genes such as NAMPT and Sirt 3. These findings indicate that NAMPT expression is a natural defense mechanism, protecting the organism against damage [57].

4.2 NAMPT functions as a growth factor

NAMPT is a pleiotropic protein that also functions via non-enzymatic mechanisms. Accumulating evidence suggests that NAMPT can function as a growth factor, which is important for regulating a large variety of cellular processes. The term “growth factor” generally refers to a naturally occurring protein capable of stimulating cellular growth, proliferation, and cellular differentiation. Conditioned medium from COS7 or PA317 cells transiently expressing hNAMPT can significantly enhance pre-B-cell colony formation of bone marrow from both human and mouse by at least 70% in the presence of both IL-7 and stem cell factor. As a result, NAMPT was initially named pre-B-cell colony enhancing factor [16]. Subsequently, NAMPT was associated with the maturation of vascular SMCs. Knockdown of endogenous NAMPT was found to increase SMC apoptosis and reduced the maturation of SMCs into a contractile state. Human SMCs transduced with the NAMPT gene exhibited enhanced survival, an elongated bipolar morphology, and increased levels of markers of a mature SMC phenotype [39].

NAMPT has also been proposed by Fukuhara and colleagues to be an adipokine (Visfatin) produced by visceral fat, which binds to and activates the insulin receptor [58]. Subsequent studies correlated plasma NAMPT levels with various metabolic states in humans, including obesity, high visceral fat mass, and diabetes [5961]. Furthermore, NAMPT was reported to have insulin mimetic effects as a growth factor for osteoblasts [62]. The effects of NAMPT on glucose uptake, proliferation, and type I collagen enhancement in cultured human osteoblast-like cells are similar to insulin and can be inhibited by a specific inhibitor of insulin receptor tyrosine kinase activity. In addition, osteocalcin secretion from human osteoblast-like cells was shown to be downregulated by NAMPT. Phosphorylation of the insulin receptor seems to be involved in this process, and the same signal transduction pathway appears to be engaged by insulin itself. However, the original publication by Fukuhara and colleagues was retracted because of issues of reproducibility of the NAMPT/insulin receptor interaction with different preparations of recombinant NAMPT protein. On the other hand, the major conclusions seem to hold true.

4.3 NAMPT acts as a proinflammatory cytokine

The first NAMPT cDNA was screened using a degenerate oligonucleotide probe designed based on the similarity of NAMPT with the coding sequences of the signal peptidase processing site in five different cytokines, including GM-CSF, IL-2, IL-1β, IL-6 and IL-13, without homology to other known cytokines [16]. A few studies indicate that NAMPT behaves like a chemokine. Lipopolysaccharide, interleukin (IL)-1beta, TNF-α and IL-6 significantly induce NAMPT expression in WISH cells, an amniotic epithelial cell line. NAMPT induction in response to these cytokines was significantly reduced in WISH cells in the presence of dexamethasone. NAMPT was also suggested to be a cytokine based on its expression in normal fetal membranes and its upregulation during infections [9]. In addition, NAMPT expression is upregulated in a variety of acute and chronic inflammatory diseases, such as sepsis [63], acute lung injury [10], rheumatoid arthritis [64], inflammatory bowel disease [65], and myocardial infarction [66]. Treatment of WISH cells and fetal membrane explants with hNAMPT significantly increases IL-6 and IL-8 gene expression, as one might expect from a proinflammatory cytokine [67].

Both extracellular and endogenous NAMPT contribute to the inhibition of neutrophil apoptosis and play a key role in the persistence of inflammation [63]. Overexpression of NAMPT significantly augments IL-8 secretion and mRNA expression in A549 cells, a human pulmonary carcinoma type II epithelial cell line and in human pulmonary artery endothelial cells. NAMPT also significantly augments IL-1β-mediated cell permeability. Furthermore, NAMPT expression affects the expression of two other inflammatory cytokines, IL-16 and CCR3 [68,69]. Hong et al. demonstrated that recombinant hNAMPT acts as a direct rat neutrophil chemotactic factor in vitro and that there is a marked increase in bronchoalveolar lavage leukocytes after intratracheal injection of hNAMPT in C57BL/6J mice [70].

4.4 Other functions of NAMPT

Genetic deletion of NAMPT is lethal in mice, indicating the essential nature of NAMPT for life [38,70]. Recently, Stein et al showed that ablation of NAMPT in excitatory neurons of the forebrain has serious repercussions for neuronal function and survival, negatively affecting cognitive function. The authors also found that CaMKIIαNAMPT−/− mice generated by crossing NAMPTflox/flox mice with CaMKIIαCre transgenic mice exhibited hyperactivity, impaired learning and memory, and reduced levels of anxiety-like behaviors, which suggest that forebrain excitatory neurons depend upon intracellular (iNAMPT)-mediated NAD biosynthesis and cannot obtain sufficient NAD+ metabolites from other sources [71]. Subsequent reports have shown that NAMPT may increase locomotion and cause orexigenic effects [72] and that NAMPT may be a serum marker of malignant astrocytoma [73]. Furthermore, NAMPT reportedly potently stimulates in vivo neovascularization in chick chorioallantoic membrane and mouse Matrigel plugs. Finally, NAMPT activates the ERK1/2 signal transduction cascades in endothelial cells, which are closely linked to angiogenesis [74].

5. Intracellular and extracellular forms of NAMPT

NAMPT exists in either the intracellular or extracellular space. iNAMPT was found in nuclei, mitochondria, and the cytosol [17]. iNAMPT participates as a rate-limiting enzyme in the salvage pathway of NAD synthesis and plays multiple roles in energy metabolism, enhancing the proliferation of endothelial cells, inhibiting apoptosis, regulating vascular tone, and stimulating autophagy in disease conditions such as stroke [51]. eNAMPT has been found both locally in non-CNS systems and circulating systemically in the blood. eNAMPT can be distinguished from iNAMPT by its larger molecular weight arising from posttranslational modifications [38]. eNAMPT, mostly in the form of serum NAMPT, likely functions as a cytokine within the peripheral circulation. eNAMPT can be released from adipocytes and participate in energy homeostasis [58]. A large number of studies have reported effects of eNAMPT on various biological functions. It is widely believed that the diverse biological functions of eNAMPT can be attributed to its NAMPT enzymatic activity. However, due to the scarcity of ATP in the extracellular space, the enzymatic activity of eNAMPT may be weak under normal circumstances [75].

6. Regional and cell-specific distribution of NAMPT in the brain

NAMPT exists ubiquitously in various tissues in human and murine brains [16,17,52]. McGlothlin et al described moderate expression of NAMPT in the brain [19]. In the study by Zhang and colleagues, the expression of NAMPT was shown to be almost exclusive to neurons in the mouse brain, absent in glial cells, and present in some endothelial cells [6]. Wang et al have shown NAMPT is expressed in both neurons and astrocytes in vitro. However, the expression of NAMPT in neurons was higher than in astrocytes, suggesting perhaps a more important role in the former cell type [7]. Liu et al showed that NAMPT levels increased in serum with aging but decreased in the aging brain in the cortex and hippocampus with no parallel change in striatum and cerebellum. Furthermore, NAMPT increased with aging in microglia but likely decreased in neurons. In young mice, immunoreactivity for NAMPT was primarily localized in NeuN-positive neurons in the cortex and hippocampal CA3 region but rarely in GFAP-positive astrocytes and Iba1-positive microglia cells. NAMPT was also found to be expressed in the Purkinje cells, granule cells, and cells in the molecular layer of cerebellum. In aged mice, NAMPT was also found to be expressed in neurons. In addition, NAMPT was highly expressed in microglial cells, but not in astrocytes [76].

When wild type hNAMPT or H247A mutant NAMPT with very weak enzymatic activity were injected intravenously into mice once every 3 d for 32 d, body weight, blood pressure, heart rate, serum glucose, serum total cholesterol and triglyceride and the morphology of neurons, astrocytes and microglia in the hippocampus were not significant affected [77]. This suggests that elevation of plasma NAMPT may not induce metabolic and neuronal dysfunction in normal individuals. In a focal cerebral infarction model, NAMPT expression was markedly increased in both the infarct core and peri-infarct penumbra, compared with the corresponding unaffected contralateral region. Additionally, NAMPT was upregulated in an in vitro model of ischemic neuronal injury from oxygen–glucose deprivation (OGD) [7].

7. Roles of NAMPT in brain ischemia

Compared with brain ischemia, the roles of NAMPT in the ischemic heart have been more widely studied. Recombinant NAMPT administration reduces myocardial injury both in vivo and in vitro, providing direct evidence for the cardioprotective effects of NAMPT. These effects were blocked by a phosphoinositide 3-kinase (PI3K) inhibitor and a MEK1/2 inhibitor [5]. Expression of NAMPT in the heart is decreased by ischemia/reperfusion and pressure overload. Cardiac-specific overexpression of NAMPT in mice increases NAD+ content in the heart, prevents downregulation of NAMPT, and reduces myocardial infarction size and apoptosis in response to prolonged ischemia and ischemia/reperfusion. These results suggest that NAMPT is an essential gatekeeper of energy status and survival in cardiac myocytes [78]. As downregulation of NAMPT impairs autophagic flux, Hsu et al suggested that NAMPT may protect against myocardial infarction by positively regulating autophagy in cardiomyocytes [78,79]. These experiments suggest that NAMPT may be an important pro-survival molecule in cardiomyocytes and that the increase of NAMPT during heart ischemia is a compensatory defense mechanism. There are many similarities between the pathophysiological mechanisms underlying ischemic heart disease and ischemic stroke. NAMPT may mediate neuroprotection by providing extra energy and activating NAD-dependent survival signaling pathways. These studies warrant further investigations of NAMPT in ischemic stroke.

As mentioned in the Introduction, stroke is a leading cause of morbidity and mortality worldwide. Ischemia leads to energy depletion and eventually neuronal death and brain damage [80,81]. NAMPT has been postulated to exert a significant impact on cellular function and survival under ischemic conditions by its multifaceted functions, including but not limited to energy metabolism. Below we review the recent literature on the complex effects of NAMPT on ischemic injury.

Lu et al reported that plasma NAMPT concentrations increased in ischemic stroke patients as an independent factor associated with ischemic stroke [82]. Similar observations were made in a mouse model [18,83]. Both eNAMPT and iNAMPT protein expression was detectable with two different NAMPT antibodies in WT C57BL/6 mice. Although iNAMPT remained unchanged, eNAMPT was dramatically induced at 24 hours after ischemia, peaking at 3 days and remaining at high levels even after 7 days. NAMPT expression was induced in both neuronal cell bodies and white matter (WM) regions, including striatal fiber bundles and the corpus callosum in WT mice after ischemia. These findings suggest that eNAMPT is selectively induced after ischemia, perhaps as part of a long-term stress response pathway.

Wang et al used lentivirus-mediated NAMPT overexpression and knockdown to manipulate NAMPT expression and explore the effects of NAMPT on neuronal survival during ischemic stress using in vivo middle cerebral artery occlusion and in vitro OGD models. Both local lentivirus-mediated NAMPT overexpression and administration of the NAMPT enzymatic product NMN reduced ischemia-induced cerebral injuries [7]. NAMPT inhibition by FK866 aggravated brain infarction after cerebral ischemia in rats. The neuroprotection exerted by NAMPT was abolished in adenosine monophosphate-activated kinase-[alpha]2 knockout (AMPK α2−/−) neurons, which supports the notion that NAMPT overexpression and knockdown regulates neuronal survival by the AMPK pathway. In neurons, NAMPT positively modulates NAD+ levels and thereby controls SIRT1 activity. SIRT1 can be co-precipitated with serine/threonine kinase 11 (LKB1), an upstream kinase of AMPK, and has been shown to promote LKB1 deacetylation in neurons, thereby controlling activation of the AMPK signaling pathway. NAMPT overexpression-induced neuroprotection was abolished in SIRT1+/− and AMPKα2−/− mice. Therefore, SIRT1 appears to be essential for NAMPT-induced AMPK activation and neuroprotection. Thus, NAMPT protects against ischemic stroke by rescuing neurons from death via the SIRT1-dependent AMPK pathway [7]. Subsequent work demonstrated that the overexpression of NAMPT increased autophagy, as indicated by LC3 expression in punctate structures, LC3-II/beclin-1 expression and increased autophagosome number both in vivo and in vitro at 2 hours after ischemic injury. At the early stages of OGD, the autophagy inducer rapamycin protected against neuronal injury induced by NAMPT knockdown, whereas the autophagy inhibitor 3-methyladenine partially abolished the neuroprotective effects of NAMPT. Overexpression or knockdown of NAMPT regulated the phosphorylation of mTOR and S6K1 signaling pathways after OGD stress by enhancing phosphorylation of TSC2 at Ser1387 but not Thr1462. Furthermore, in cultured SIRT1-knockout neurons, the regulation of autophagic proteins LC3-II and beclin-1 by NAMPT was abolished. These results reveal that NAMPT promotes neuronal by promoting autophagy through the TSC2-mTOR-S6K1 signaling pathway in a SIRT1-dependent manner during cerebral ischemia.

Heterozygote NAMPT-knockout (NAMPT+/−) mice with reduced expression of NAMPT have been used to determine the role of NAMPT in cerebral ischemia induced by photothrombosis. NAMPT+/− mice exhibit a higher density of degenerating, Fluoro-Jade B+ neurons in the penumbra than WT mice. These results provide evidence that neuronal NAMPT is protective against cerebral ischemia [6]. In vitro OGD and glutamate excitotoxicity models have been used to study the mechanism underlying the protective effects of NAMPT on primary neurons. Treatment of primary neurons with NAM and NAD+ significantly reduces neuronal death after OGD and glutamate excitotoxicity, respectively, whereas treatment of neurons with FK866 increases neuronal death after OGD. Furthermore, over-expression of hNAMPT reduces glutamate excitotoxicity, whereas over-expression of hNAMPT mutants lacking enzymatic activity exerts no effect on neuronal death. In addition, the addition of NAD+ and NAM increases mitochondrial biogenesis in neurons after OGD. Over-expression of NAMPT in neurons reduces mitochondrial membrane potential depolarization following glutamate stimulation, whereas over-expression of hNAMPT mutants without enzymatic activity does not affect mitochondrial membrane potential depolarization. Thus, NAMPT exerts neuroprotective effects in ischemia via enzymatic activity for NAD+ production and amelioration of mitochondrial dysfunction [84].

Zhao et al demonstrated that plasma concentrations of NAMPT, NAD+, and ATP were all increased in mice after cerebral ischemia [83]. For the first time, they reported that cultured glia, but not neurons, increased their secretion of NAMPT under OGD stress conditions, as shown by immunoblotting and enzyme immunoassays. Treatment with wild-type NAMPT, but not H247A-mutant enzymatic-dead NAMPT, significantly attenuated OGD-induced cell death and apoptosis in both cultured mouse neurons and glia. Conversely, treatment with neutralizing antibodies abolished the protective effects of eNAMPT on cell viability, although this treatment failed to block the anti-apoptotic effects of eNAMPT [83]. Furthermore, plasma NAMPT concentrations were decreased in 6-month-old but not 3-month-old spontaneously hypertensive stroke-prone (SHR-SP) rats, a spontaneous stroke animal model, compared with age-matched Wistar-Kyoto rats. The decrease in plasma visfatin levels in 6-month-old SHR-SP rats is correlated with the onset of susceptibility to stroke at around 6-months of age [85]. More importantly, treatment with FK866 for 3 months does not induce death or abnormal behavior in normal rats, although inhibition of NAMPT enzymatic function by FK866 accelerates stroke occurrence and death in SHR-SP rats [83]. The authors of these studies argue that eNAMPT exhibits enzymatic activity and may be as neuroprotective as iNAMPT in cerebral ischemic injury.

Cerebral ischemic injury leads to both gray matter and WM pathology. WM is highly vulnerable to ischemia and often more severely injured than gray matter [86]. In most cases of human stroke, WM injury accounts for half of the lesion volume [87]. Despite the essential roles of WM in neural function and the susceptibility of WM to ischemia, WM injury has been largely overlooked in animal studies as well as in clinical treatments [88]. However, complete neuroprotection cannot be attained without WM protection. NAMPT can delay axon degeneration in the presence of NAM in an in vitro Wallerian degeneration assay [89]. Sasaki et al. suggest that stimulation of NAD biosynthetic pathways by a variety of interventions may be useful in preventing or delaying axonal degeneration [89]. Our group studied the role of NAMPT in both gray and white matter protection against ischemia in vivo. We confirmed the protective effects of NAMPT against ischemic stroke induced in mice by transient occlusion of the middle cerebral artery. Neuron-specific NAMPT overexpression significantly reduced infarct volume and improved long-term neurologic outcomes compared with littermates. Interestingly, neuronal overexpression of NAMPT increased the area of myelinated fibers in the striatum and corpus callosum, indicating that NAMPT protects against WM injury. We then provided evidence that the protective effects are mediated by eNAMPT. Conditioned medium from NAMPT-overexpressing neurons exposed to OGD protected cultured oligodendrocytes from OGD and these effects were abolished with neutralizing antibodies [18]. Thus, we concluded that NAMPT may not function simply in a restricted intracellular manner during stroke recovery but that it protects both gray matter and WM from stroke injury through extracellular actions.

In sum, most studies convincingly support a protective role for iNAMPT against brain injury. However, there is less agreement on the effects of eNAMPT on ischemic injury. In contrast with the studies mentioned above, which indicate protective roles of NAMPT against cerebral ischemic injury, Zhao et al reported that cerebral ischemic-like injury in cultured neural cells was exacerbated by eNAMPT. Furthermore, administration of recombinant NAMPT protein exacerbated MCAO-induced neuronal injury in rat brain. NAMPT exacerbated OGD-induced neuronal injury only in mixed neuron-glia cultures, but not in pure neuron cultures. In the mixed cultures, NAMPT protein promoted TNF-α release in a time- and concentration-dependent fashion. Conversely TNF-α neutralizing antibodies protected against the enhancement of OGD neuronal injury by NAMPT. In addition, the H247A mutant of NAMPT lacking all enzymatic activity exerted similar effects on ischemic neuronal injury and TNF-α release as the wild type protein. Thus, the authors concluded that eNAMPT is an injurious and inflammatory factor in cerebral ischemia and aggravates ischemic neuronal injury by triggering TNF-α release from glial cells, through a mechanism unrelated to NAMPT enzymatic activity [8].

8. More considerations THAT may affect the role of NAMPT in ischemic brain

The precise role of NAMPT in cerebrovascular diseases, including ischemic brain injury, needs to be fully elucidated because of its controversial biological actions on a variety of cellular functions. Ischemic stroke damages many different types of cells in the brain aside from neurons and glia. For example, dysfunction in macrophages, vascular smooth muscle cells (VSMCs), and endothelial cells is believed to to underlie the genesis and development of atherosclerosis, and it is generally accepted that NAMPT is a proinflammatory factor in atherosclerosis. However, NAMPT is not only an inflammatory cytokine that promotes endothelial cell activation, VSMCs proliferation, and macrophage migration, but also a key protector of the same cells against various stresses [37,40,54,90,91]. These complex features of NAMPT are consistent with the evolution of cytokines as part of an adaptive immune response to injury. Inflammatory mediators affect the integrity of the neurovascular unit, including the endothelium, astrocytes, the vascular extracellular matrix, pericytes, and neurons and their axons. Indeed, all components of the neurovascular unit respond to inflammation, which is known to be initiated by ischemia. Under ischemic conditions, pericytes constrict brain capillaries and then die, which may lead to a long-lasting decrease in blood flow and loss of blood–brain barrier function, thereby increasing death of nerve cells [92]. However, how other components of the neurovascular unit communicate during post-ischemic inflammatory responses is not yet clear, as a unifying scheme of inflammatory responses has not yet been developed. As such, the effects of NAMPT on cellular processes may be double-edged and depend on its expression level and distribution. Thus, many issues remain unsolved, although studies in various tissues have provided valuable information on the relationship between NAMPT and ischemic stroke and improved our understanding of the underlying mechanisms.

9. Concluding Remarks

NAMPT has drawn much attention in biomedical fields because of its pleiotropic physiological functions and because its dysregulation is implicated in a number of human diseases, including ischemia. Recent studies have provided new insights into the molecular effects of NAMPT using the specific inhibitor FK866 and genetic means of manipulating NAMPT expression. Collectively, the majority of experimental studies conducted thus far suggest that NAMPT is an attractive therapeutic target for both the prevention and treatment of ischemic stroke. Zhao et al have commented on some discrepancies between studies, suggesting that the duration of OGD may be critical [83]. In addition, standardized procedures for the preparation of NAMPT protein constructs are urgently needed. Further exploration is needed of the potential effects of trace amounts of residual endotoxins from E.Coli in recombinant preparations of NAMPT. Moreover, some reports have claimed that NAMPT is a ligand for the insulin receptor [28,58]. However, the actual receptor for NAMPT has not yet been convincingly identified. The mechanisms underlying the cytokine-like effects of NAMPT also await further exploration. It is worth noting that the extracellular mode of action of NAMPT and its ability to cross the blood-brain barrier both make it a clinically viable candidate for stroke therapy. In conclusion, further investigations into the properties of NAMPT are highly warranted.

Acknowledgments

We thank all the authors who contributed to this field of work and apologize to those whose works are not directly cited in this review. This work was supported in part by NIH grants R01 NS079345 and VA Merit Review grants RX000199/BX002346 (Dr. Cao).

References

  • 1.Gibson CL, Bath PM, Murphy SP. G-CSF reduces infarct volume improves functional outcome after transient focal cerebral ischemia in mice. J Cereb Blood Flow Metab. 2005;25(4):431–439. doi: 10.1038/sj.jcbfm.9600033. [DOI] [PubMed] [Google Scholar]
  • 2.Eliasson MJ, Sampei K, Mandir AS, Hurn PD, Traystman RJ, Bao J, Pieper A, Wang ZQ, Dawson TM, Snyder SH, Dawson VL. Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med. 1997;3(10):1089–1095. doi: 10.1038/nm1097-1089. [DOI] [PubMed] [Google Scholar]
  • 3.Endres M, Wang ZQ, Namura S, Waeber C, Moskowitz MA. Ischemic brain injury is mediated by the activation of poly(ADP-ribose)polymerase. J Cereb Blood Flow Metab. 1997;17(11):1143–1151. doi: 10.1097/00004647-199711000-00002. [DOI] [PubMed] [Google Scholar]
  • 4.Yu SW, Wang H, Dawson TM, Dawson VL. Poly(ADP-ribose) polymerase-1 and apoptosis inducing factor in neurotoxicity. Neurobiol Dis. 2003;14(3):303–317. doi: 10.1016/j.nbd.2003.08.008. [DOI] [PubMed] [Google Scholar]
  • 5.Lim SY, Davidson SM, Paramanathan AJ, Smith CC, Yellon DM, Hausenloy DJ. The novel adipocytokine visfatin exerts direct cardioprotective effects. J Cell Mol Med. 2008;12(4):1395–1403. doi: 10.1111/j.1582-4934.2008.00332.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhang W, Xie Y, Wang T, Bi J, Li H, Zhang LQ, Ye SQ, Ding S. Neuronal protective role of PBEF in a mouse model of cerebral ischemia. J Cereb Blood Flow Metab. 2010;30(12):1962–1971. doi: 10.1038/jcbfm.2010.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wang P, Xu TY, Guan YF, Tian WW, Viollet B, Rui YC, Zhai QW, Su DF, Miao CY. Nicotinamide phosphoribosyltransferase protects against ischemic stroke through SIRT1-dependent adenosine monophosphate-activated kinase pathway. Ann Neurol. 2011;69(2):360–374. doi: 10.1002/ana.22236. [DOI] [PubMed] [Google Scholar]
  • 8.Zhao B, Zhang M, Han X, Zhang XY, Xing Q, Dong X, Shi QJ, Huang P, Lu YB, Wei EQ, Xia Q, Zhang WP, Tang C. Cerebral ischemia is exacerbated by extracellular nicotinamide phosphoribosyltransferase via a non-enzymatic mechanism. PLoS One. 2013;8(12):e85403. doi: 10.1371/journal.pone.0085403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ognjanovic S, Bao S, Yamamoto SY, Garibay-Tupas J, Samal B, Bryant-Greenwood GD. Genomic organization of the gene coding for human pre-B-cell colony enhancing factor and expression in human fetal membranes. J Mol Endocrinol. 2001;26(2):107–117. doi: 10.1677/jme.0.0260107. [DOI] [PubMed] [Google Scholar]
  • 10.Ye SQ, Simon BA, Maloney JP, Zambelli-Weiner A, Gao L, Grant A, Easley RB, McVerry BJ, Tuder RM, Standiford T, Brower RG, Barnes KC, Garcia JG. Pre-B-cell colony-enhancing factor as a potential novel biomarker in acute lung injury. Am J Respir Crit Care Med. 2005;171(4):361–370. doi: 10.1164/rccm.200404-563OC. [DOI] [PubMed] [Google Scholar]
  • 11.Bajwa EK, Yu CL, Gong MN, Thompson BT, Christiani DC. Pre-B-cell colony-enhancing factor gene polymorphisms and risk of acute respiratory distress syndrome. Crit Care Med. 2007;35(5):1290–1295. doi: 10.1097/01.CCM.0000260243.22758.4F. [DOI] [PubMed] [Google Scholar]
  • 12.Johansson LM, Johansson LE, Ridderstrale M. The visfatin (PBEF1) G-948T gene polymorphism is associated with increased high-density lipoprotein cholesterol in obese subjects. Metabolism. 2008;57(11):1558–1562. doi: 10.1016/j.metabol.2008.06.011. [DOI] [PubMed] [Google Scholar]
  • 13.Tokunaga A, Miura A, Okauchi Y, Segawa K, Fukuhara A, Okita K, Takahashi M, Funahashi T, Miyagawa J, Shimomura I, Yamagata K. The -1535 promoter variant of the visfatin gene is associated with serum triglyceride and HDL-cholesterol levels in Japanese subjects. Endocr J. 2008;55(1):205–212. doi: 10.1507/endocrj.k07e-039. [DOI] [PubMed] [Google Scholar]
  • 14.Bailey SD, Loredo-Osti JC, Lepage P, Faith J, Fontaine J, Desbiens KM, Hudson TJ, Bouchard C, Gaudet D, Perusse L, Vohl MC, Engert JC. Common polymorphisms in the promoter of the visfatin gene (PBEF1) influence plasma insulin levels in a French-Canadian population. Diabetes. 2006;55(10):2896–2902. doi: 10.2337/db06-0189. [DOI] [PubMed] [Google Scholar]
  • 15.Bottcher Y, Teupser D, Enigk B, Berndt J, Kloting N, Schon MR, Thiery J, Bluher M, Stumvoll M, Kovacs P. Genetic variation in the visfatin gene (PBEF1) and its relation to glucose metabolism and fat-depot-specific messenger ribonucleic acid expression in humans. J Clin Endocrinol Metab. 2006;91(7):2725–2731. doi: 10.1210/jc.2006-0149. [DOI] [PubMed] [Google Scholar]
  • 16.Samal B, Sun Y, Stearns G, Xie C, Suggs S, McNiece I. Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol Cell Biol. 1994;14(2):1431–1437. doi: 10.1128/mcb.14.2.1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kitani T, Okuno S, Fujisawa H. Growth phase-dependent changes in the subcellular localization of pre-B-cell colony-enhancing factor. FEBS Lett. 2003;544(1–3):74–78. doi: 10.1016/s0014-5793(03)00476-9. [DOI] [PubMed] [Google Scholar]
  • 18.Jing Z, Xing J, Chen X, Stetler RA, Weng Z, Gan Y, Zhang F, Gao Y, Chen J, Leak RK, Cao G. Neuronal NAMPT is released after cerebral ischemia and protects against white matter injury. J Cereb Blood Flow Metab. 2014;34(10):1613–1621. doi: 10.1038/jcbfm.2014.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.McGlothlin JR, Gao L, Lavoie T, Simon BA, Easley RB, Ma SF, Rumala BB, Garcia JG, Ye SQ. Molecular cloning and characterization of canine pre-B-cell colony-enhancing factor. Biochem Genet. 2005;43(3–4):127–141. doi: 10.1007/s10528-005-1505-2. [DOI] [PubMed] [Google Scholar]
  • 20.Rongvaux A, Shea RJ, Mulks MH, Gigot D, Urbain J, Leo O, Andris F. Pre-B-cell colony-enhancing factor, whose expression is up-regulated in activated lymphocytes, is a nicotinamide phosphoribosyltransferase, a cytosolic enzyme involved in NAD biosynthesis. Eur J Immunol. 2002;32(11):3225–3234. doi: 10.1002/1521-4141(200211)32:11<3225::AID-IMMU3225>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 21.Kang GB, Bae MH, Kim MK, Im I, Kim YC, Eom SH. Crystal structure of Rattus norvegicus Visfatin/PBEF/Nampt in complex with an FK866-based inhibitor. Mol Cells. 2009;27(6):667–671. doi: 10.1007/s10059-009-0088-x. [DOI] [PubMed] [Google Scholar]
  • 22.Khan JA, Tao X, Tong L. Molecular basis for the inhibition of human NMPRTase, a novel target for anticancer agents. Nat Struct Mol Biol. 2006;13(7):582–588. doi: 10.1038/nsmb1105. [DOI] [PubMed] [Google Scholar]
  • 23.Gross JW, Rajavel M, Grubmeyer C. Kinetic mechanism of nicotinic acid phosphoribosyltransferase: implications for energy coupling. Biochemistry. 1998;37(12):4189–4199. doi: 10.1021/bi972014w. [DOI] [PubMed] [Google Scholar]
  • 24.Wang T, Zhang X, Bheda P, Revollo JR, Imai S, Wolberger C. Structure of Nampt/PBEF/visfatin, a mammalian NAD+ biosynthetic enzyme. Nat Struct Mol Biol. 2006;13(7):661–662. doi: 10.1038/nsmb1114. [DOI] [PubMed] [Google Scholar]
  • 25.Tanaka M, Nozaki M, Fukuhara A, Segawa K, Aoki N, Matsuda M, Komuro R, Shimomura I. Visfatin is released from 3T3-L1 adipocytes via a non-classical pathway. Biochem Biophys Res Commun. 2007;359(2):194–201. doi: 10.1016/j.bbrc.2007.05.096. [DOI] [PubMed] [Google Scholar]
  • 26.Garten A, Petzold S, Barnikol-Oettler A, Korner A, Thasler WE, Kratzsch J, Kiess W, Gebhardt R. Nicotinamide phosphoribosyltransferase (NAMPT/PBEF/visfatin) is constitutively released from human hepatocytes. Biochem Biophys Res Commun. 2010;391(1):376–381. doi: 10.1016/j.bbrc.2009.11.066. [DOI] [PubMed] [Google Scholar]
  • 27.Hasmann M, Schemainda I. FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. Cancer Res. 2003;63(21):7436–7442. [PubMed] [Google Scholar]
  • 28.Kim MK, Lee JH, Kim H, Park SJ, Kim SH, Kang GB, Lee YS, Kim JB, Kim KK, Suh SW, Eom SH. Crystal structure of visfatin/pre-B cell colony-enhancing factor 1/nicotinamide phosphoribosyltransferase, free and in complex with the anti-cancer agent FK-866. J Mol Biol. 2006;362(1):66–77. doi: 10.1016/j.jmb.2006.06.082. [DOI] [PubMed] [Google Scholar]
  • 29.Nahimana A, Attinger A, Aubry D, Greaney P, Ireson C, Thougaard AV, Tjornelund J, Dawson KM, Dupuis M, Duchosal MA. The NAD biosynthesis inhibitor APO866 has potent antitumor activity against hematologic malignancies. Blood. 2009;113(14):3276–3286. doi: 10.1182/blood-2008-08-173369. [DOI] [PubMed] [Google Scholar]
  • 30.Zoppoli G, Cea M, Soncini D, Fruscione F, Rudner J, Moran E, Caffa I, Bedognetti D, Motta G, Ghio R, Ferrando F, Ballestrero A, Parodi S, Belka C, Patrone F, Bruzzone S, Nencioni A. Potent synergistic interaction between the Nampt inhibitor APO866 and the apoptosis activator TRAIL in human leukemia cells. Exp Hematol. 2010;38(11):979–988. doi: 10.1016/j.exphem.2010.07.013. [DOI] [PubMed] [Google Scholar]
  • 31.Hjarnaa PJ, Jonsson E, Latini S, Dhar S, Larsson R, Bramm E, Skov T, Binderup L. CHS 828, a novel pyridyl cyanoguanidine with potent antitumor activity in vitro and in vivo. Cancer Res. 1999;59(22):5751–5757. [PubMed] [Google Scholar]
  • 32.Zheng X, Baumeister T, Buckmelter AJ, Caligiuri M, Clodfelter KH, Han B, Ho YC, Kley N, Lin J, Reynolds DJ, Sharma G, Smith CC, Wang Z, Dragovich PS, Oh A, Wang W, Zak M, Wang Y, Yuen PW, Bair KW. Discovery of potent and efficacious cyanoguanidine-containing nicotinamide phosphoribosyltransferase (Nampt) inhibitors. Bioorg Med Chem Lett. 2014;24(1):337–343. doi: 10.1016/j.bmcl.2013.11.006. [DOI] [PubMed] [Google Scholar]
  • 33.Olesen UH, Thougaard AV, Jensen PB, Sehested M. A preclinical study on the rescue of normal tissue by nicotinic acid in high-dose treatment with APO866, a specific nicotinamide phosphoribosyltransferase inhibitor. Mol Cancer Ther. 2010;9(6):1609–1617. doi: 10.1158/1535-7163.MCT-09-1130. [DOI] [PubMed] [Google Scholar]
  • 34.von Heideman A, Berglund A, Larsson R, Nygren P. Safety and efficacy of NAD depleting cancer drugs: results of a phase I clinical trial of CHS 828 and overview of published data. Cancer Chemother Pharmacol. 2010;65(6):1165–1172. doi: 10.1007/s00280-009-1125-3. [DOI] [PubMed] [Google Scholar]
  • 35.Olesen UH, Christensen MK, Bjorkling F, Jaattela M, Jensen PB, Sehested M, Nielsen SJ. Anticancer agent CHS-828 inhibits cellular synthesis of NAD. Biochem Biophys Res Commun. 2008;367(4):799–804. doi: 10.1016/j.bbrc.2008.01.019. [DOI] [PubMed] [Google Scholar]
  • 36.Olesen UH, Petersen JG, Garten A, Kiess W, Yoshino J, Imai S, Christensen MK, Fristrup P, Thougaard AV, Bjorkling F, Jensen PB, Nielsen SJ, Sehested M. Target enzyme mutations are the molecular basis for resistance towards pharmacological inhibition of nicotinamide phosphoribosyltransferase. BMC Cancer. 2010;10:677. doi: 10.1186/1471-2407-10-677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wang P, Xu TY, Guan YF, Su DF, Fan GR, Miao CY. Perivascular adipose tissue-derived visfatin is a vascular smooth muscle cell growth factor: role of nicotinamide mononucleotide. Cardiovasc Res. 2009;81(2):370–380. doi: 10.1093/cvr/cvn288. [DOI] [PubMed] [Google Scholar]
  • 38.Revollo JR, Korner A, Mills KF, Satoh A, Wang T, Garten A, Dasgupta B, Sasaki Y, Wolberger C, Townsend RR, Milbrandt J, Kiess W, Imai S. Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab. 2007;6(5):363–375. doi: 10.1016/j.cmet.2007.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.van der Veer E, Nong Z, O’Neil C, Urquhart B, Freeman D, Pickering JG. Pre-B-cell colony-enhancing factor regulates NAD+-dependent protein deacetylase activity and promotes vascular smooth muscle cell maturation. Circ Res. 2005;97(1):25–34. doi: 10.1161/01.RES.0000173298.38808.27. [DOI] [PubMed] [Google Scholar]
  • 40.Borradaile NM, Pickering JG. Nicotinamide phosphoribosyltransferase imparts human endothelial cells with extended replicative lifespan and enhanced angiogenic capacity in a high glucose environment. Aging Cell. 2009;8(2):100–112. doi: 10.1111/j.1474-9726.2009.00453.x. [DOI] [PubMed] [Google Scholar]
  • 41.Corda D, Di Girolamo M. Functional aspects of protein mono-ADP-ribosylation. EMBO J. 2003;22(9):1953–1958. doi: 10.1093/emboj/cdg209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Menissier de Murcia J, Ricoul M, Tartier L, Niedergang C, Huber A, Dantzer F, Schreiber V, Ame JC, Dierich A, LeMeur M, Sabatier L, Chambon P, de Murcia G. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J. 2003;22(9):2255–2263. doi: 10.1093/emboj/cdg206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lee HC. Physiological functions of cyclic ADP-ribose and NAADP as calcium messengers. Annu Rev Pharmacol Toxicol. 2001;41:317–345. doi: 10.1146/annurev.pharmtox.41.1.317. [DOI] [PubMed] [Google Scholar]
  • 44.Blander G, Guarente L. The Sir2 family of protein deacetylases. Annu Rev Biochem. 2004;73:417–435. doi: 10.1146/annurev.biochem.73.011303.073651. [DOI] [PubMed] [Google Scholar]
  • 45.Campisi J. Aging, chromatin, and food restriction–connecting the dots. Science. 2000;289(5487):2062–2063. doi: 10.1126/science.289.5487.2062. [DOI] [PubMed] [Google Scholar]
  • 46.Schreiber V, Dantzer F, Ame JC, de Murcia G. Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol. 2006;7(7):517–528. doi: 10.1038/nrm1963. [DOI] [PubMed] [Google Scholar]
  • 47.Wright SC, Wei QS, Kinder DH, Larrick JW. Biochemical pathways of apoptosis: nicotinamide adenine dinucleotide-deficient cells are resistant to tumor necrosis factor or ultraviolet light activation of the 24-kD apoptotic protease and DNA fragmentation. J Exp Med. 1996;183(2):463–471. doi: 10.1084/jem.183.2.463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Berger F, Ramirez-Hernandez MH, Ziegler M. The new life of a centenarian: signalling functions of NAD(P) Trends Biochem Sci. 2004;29(3):111–118. doi: 10.1016/j.tibs.2004.01.007. [DOI] [PubMed] [Google Scholar]
  • 49.Ziegler M. New functions of a long-known molecule. Emerging roles of NAD in cellular signaling. Eur J Biochem. 2000;267(6):1550–1564. doi: 10.1046/j.1432-1327.2000.01187.x. [DOI] [PubMed] [Google Scholar]
  • 50.Belenky P, Bogan KL, Brenner C. NAD+ metabolism in health and disease. Trends Biochem Sci. 2007;32(1):12–19. doi: 10.1016/j.tibs.2006.11.006. [DOI] [PubMed] [Google Scholar]
  • 51.Garten A, Petzold S, Korner A, Imai S, Kiess W. Nampt: linking NAD biology, metabolism and cancer. Trends Endocrinol Metab. 2009;20(3):130–138. doi: 10.1016/j.tem.2008.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Martin PR, Shea RJ, Mulks MH. Identification of a plasmid-encoded gene from Haemophilus ducreyi which confers NAD independence. J Bacteriol. 2001;183(4):1168–1174. doi: 10.1128/JB.183.4.1168-1174.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279(49):50754–50763. doi: 10.1074/jbc.M408388200. [DOI] [PubMed] [Google Scholar]
  • 54.van der Veer E, Ho C, O’Neil C, Barbosa N, Scott R, Cregan SP, Pickering JG. Extension of human cell lifespan by nicotinamide phosphoribosyltransferase. J Biol Chem. 2007;282(15):10841–10845. doi: 10.1074/jbc.C700018200. [DOI] [PubMed] [Google Scholar]
  • 55.Yang H, Yang T, Baur JA, Perez E, Matsui T, Carmona JJ, Lamming DW, Souza-Pinto NC, Bohr VA, Rosenzweig A, de Cabo R, Sauve AA, Sinclair DA. Nutrient-sensitive mitochondrial NAD+ levels dictate cell survival. Cell. 2007;130(6):1095–1107. doi: 10.1016/j.cell.2007.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Adya R, Tan BK, Punn A, Chen J, Randeva HS. Visfatin induces human endothelial VEGF and MMP-2/9 production via MAPK and PI3K/Akt signalling pathways: novel insights into visfatin-induced angiogenesis. Cardiovasc Res. 2008;78(2):356–365. doi: 10.1093/cvr/cvm111. [DOI] [PubMed] [Google Scholar]
  • 57.Benigni A, Corna D, Zoja C, Sonzogni A, Latini R, Salio M, Conti S, Rottoli D, Longaretti L, Cassis P, Morigi M, Coffman TM, Remuzzi G. Disruption of the Ang II type 1 receptor promotes longevity in mice. J Clin Invest. 2009;119(3):524–530. doi: 10.1172/JCI36703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M, Kishimoto K, Matsuki Y, Murakami M, Ichisaka T, Murakami H, Watanabe E, Takagi T, Akiyoshi M, Ohtsubo T, Kihara S, Yamashita S, Makishima M, Funahashi T, Yamanaka S, Hiramatsu R, Matsuzawa Y, Shimomura I. Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science. 2005;307(5708):426–430. doi: 10.1126/science.1097243. [DOI] [PubMed] [Google Scholar]
  • 59.Haider DG, Schindler K, Schaller G, Prager G, Wolzt M, Ludvik B. Increased plasma visfatin concentrations in morbidly obese subjects are reduced after gastric banding. J Clin Endocrinol Metab. 2006;91(4):1578–1581. doi: 10.1210/jc.2005-2248. [DOI] [PubMed] [Google Scholar]
  • 60.Sandeep S, Velmurugan K, Deepa R, Mohan V. Serum visfatin in relation to visceral fat, obesity, and type 2 diabetes mellitus in Asian Indians. Metabolism. 2007;56(4):565–570. doi: 10.1016/j.metabol.2006.12.005. [DOI] [PubMed] [Google Scholar]
  • 61.Chen MP, Chung FM, Chang DM, Tsai JC, Huang HF, Shin SJ, Lee YJ. Elevated plasma level of visfatin/pre-B cell colony-enhancing factor in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2006;91(1):295–299. doi: 10.1210/jc.2005-1475. [DOI] [PubMed] [Google Scholar]
  • 62.Xie H, Tang SY, Luo XH, Huang J, Cui RR, Yuan LQ, Zhou HD, Wu XP, Liao EY. Insulin-like effects of visfatin on human osteoblasts. Calcif Tissue Int. 2007;80(3):201–210. doi: 10.1007/s00223-006-0155-7. [DOI] [PubMed] [Google Scholar]
  • 63.Jia SH, Li Y, Parodo J, Kapus A, Fan L, Rotstein OD, Marshall JC. Pre-B cell colony-enhancing factor inhibits neutrophil apoptosis in experimental inflammation and clinical sepsis. J Clin Invest. 2004;113(9):1318–1327. doi: 10.1172/JCI19930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Brentano F, Schorr O, Ospelt C, Stanczyk J, Gay RE, Gay S, Kyburz D. Pre-B cell colony-enhancing factor/visfatin, a new marker of inflammation in rheumatoid arthritis with proinflammatory and matrix-degrading activities. Arthritis Rheum. 2007;56(9):2829–2839. doi: 10.1002/art.22833. [DOI] [PubMed] [Google Scholar]
  • 65.Tilg H, Moschen AR. Role of adiponectin and PBEF/visfatin as regulators of inflammation: involvement in obesity-associated diseases. Clin Sci (Lond) 2008;114(4):275–288. doi: 10.1042/CS20070196. [DOI] [PubMed] [Google Scholar]
  • 66.Dahl TB, Yndestad A, Skjelland M, Oie E, Dahl A, Michelsen A, Damas JK, Tunheim SH, Ueland T, Smith C, Bendz B, Tonstad S, Gullestad L, Froland SS, Krohg-Sorensen K, Russell D, Aukrust P, Halvorsen B. Increased expression of visfatin in macrophages of human unstable carotid and coronary atherosclerosis: possible role in inflammation and plaque destabilization. Circulation. 2007;115(8):972–980. doi: 10.1161/CIRCULATIONAHA.106.665893. [DOI] [PubMed] [Google Scholar]
  • 67.Ognjanovic S, Bryant-Greenwood GD. Pre-B-cell colony-enhancing factor, a novel cytokine of human fetal membranes. Am J Obstet Gynecol. 2002;187(4):1051–1058. doi: 10.1067/mob.2002.126295. [DOI] [PubMed] [Google Scholar]
  • 68.Li H, Liu P, Cepeda J, Fang D, Easley RB, Simon BA, Zhang LQ, Ye SQ. Augmentation of Pulmonary Epithelial Cell IL-8 Expression and Permeability by Pre-B-cell Colony Enhancing Factor. J Inflamm (Lond) 2008;5:15. doi: 10.1186/1476-9255-5-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liu P, Li H, Cepeda J, Zhang LQ, Cui X, Garcia JG, Ye SQ. Critical role of PBEF expression in pulmonary cell inflammation and permeability. Cell Biol Int. 2009;33(1):19–30. doi: 10.1016/j.cellbi.2008.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hong SB, Huang Y, Moreno-Vinasco L, Sammani S, Moitra J, Barnard JW, Ma SF, Mirzapoiazova T, Evenoski C, Reeves RR, Chiang ET, Lang GD, Husain AN, Dudek SM, Jacobson JR, Ye SQ, Lussier YA, Garcia JG. Essential role of pre-B-cell colony enhancing factor in ventilator-induced lung injury. Am J Respir Crit Care Med. 2008;178(6):605–617. doi: 10.1164/rccm.200712-1822OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Stein LR, Wozniak DF, Dearborn JT, Kubota S, Apte RS, Izumi Y, Zorumski CF, Imai S. Expression of Nampt in hippocampal and cortical excitatory neurons is critical for cognitive function. J Neurosci. 2014;34(17):5800–5815. doi: 10.1523/JNEUROSCI.4730-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Cline MA, Nandar W, Prall BC, Bowden CN, Denbow DM. Central visfatin causes orexigenic effects in chicks. Behav Brain Res. 2008;186(2):293–297. doi: 10.1016/j.bbr.2007.08.016. [DOI] [PubMed] [Google Scholar]
  • 73.Reddy PS, Umesh S, Thota B, Tandon A, Pandey P, Hegde AS, Balasubramaniam A, Chandramouli BA, Santosh V, Rao MR, Kondaiah P, Somasundaram K. PBEF1/NAmPRTase/Visfatin: a potential malignant astrocytoma/glioblastoma serum marker with prognostic value. Cancer Biol Ther. 2008;7(5):663–668. doi: 10.4161/cbt.7.5.5663. [DOI] [PubMed] [Google Scholar]
  • 74.Kim SR, Bae SK, Choi KS, Park SY, Jun HO, Lee JY, Jang HO, Yun I, Yoon KH, Kim YJ, Yoo MA, Kim KW, Bae MK. Visfatin promotes angiogenesis by activation of extracellular signal-regulated kinase 1/2. Biochem Biophys Res Commun. 2007;357(1):150–156. doi: 10.1016/j.bbrc.2007.03.105. [DOI] [PubMed] [Google Scholar]
  • 75.Hara N, Yamada K, Shibata T, Osago H, Tsuchiya M. Nicotinamide phosphoribosyltransferase/visfatin does not catalyze nicotinamide mononucleotide formation in blood plasma. PLoS One. 2011;6(8):e22781. doi: 10.1371/journal.pone.0022781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liu LY, Wang F, Zhang XY, Huang P, Lu YB, Wei EQ, Zhang WP. Nicotinamide phosphoribosyltransferase may be involved in age-related brain diseases. PLoS One. 2012;7(10):e44933. doi: 10.1371/journal.pone.0044933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yuan Q, Zhang W. Effects of recombinant human NAMPT on physiological/biochemical indexes and brain structure in mice. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2013;42(3):303–310. [PubMed] [Google Scholar]
  • 78.Hsu CP, Oka S, Shao D, Hariharan N, Sadoshima J. Nicotinamide phosphoribosyltransferase regulates cell survival through NAD+ synthesis in cardiac myocytes. Circ Res. 2009;105(5):481–491. doi: 10.1161/CIRCRESAHA.109.203703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hsu CP, Hariharan N, Alcendor RR, Oka S, Sadoshima J. Nicotinamide phosphoribosyltransferase regulates cell survival through autophagy in cardiomyocytes. Autophagy. 2009;5(8):1229–1231. doi: 10.4161/auto.5.8.10275. [DOI] [PubMed] [Google Scholar]
  • 80.Dirnagl U, Iadecola C, Moskowitz MA. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci. 1999;22(9):391–397. doi: 10.1016/s0166-2236(99)01401-0. [DOI] [PubMed] [Google Scholar]
  • 81.Lo EH, Dalkara T, Moskowitz MA. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 2003;4(5):399–415. doi: 10.1038/nrn1106. [DOI] [PubMed] [Google Scholar]
  • 82.Lu LF, Yang SS, Wang CP, Hung WC, Yu TH, Chiu CA, Chung FM, Shin SJ, Lee YJ. Elevated visfatin/pre-B-cell colony-enhancing factor plasma concentration in ischemic stroke. J Stroke Cerebrovasc Dis. 2009;18(5):354–359. doi: 10.1016/j.jstrokecerebrovasdis.2009.01.003. [DOI] [PubMed] [Google Scholar]
  • 83.Zhao Y, Liu XZ, Tian WW, Guan YF, Wang P, Miao CY. Extracellular visfatin has nicotinamide phosphoribosyltransferase enzymatic activity and is neuroprotective against ischemic injury. CNS Neurosci Ther. 2014;20(6):539–547. doi: 10.1111/cns.12273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bi J, Li H, Ye SQ, Ding S. Pre-B-cell colony-enhancing factor exerts a neuronal protection through its enzymatic activity and the reduction of mitochondrial dysfunction in in vitro ischemic models. J Neurochem. 2012;120(2):334–346. doi: 10.1111/j.1471-4159.2011.07566.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang P, Tian WW, Song J, Guan YF, Miao CY. Deficiency of NG2+ cells contributes to the susceptibility of stroke-prone spontaneously hypertensive rats. CNS Neurosci Ther. 2011;17(5):327–332. doi: 10.1111/j.1755-5949.2011.00265.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pantoni L, Garcia JH, Gutierrez JA. Cerebral white matter is highly vulnerable to ischemia. Stroke. 1996;27(9):1641–1646. doi: 10.1161/01.str.27.9.1641. discussion 1647. [DOI] [PubMed] [Google Scholar]
  • 87.Ho PW, Reutens DC, Phan TG, Wright PM, Markus R, Indra I, Young D, Donnan GA. Is white matter involved in patients entered into typical trials of neuroprotection? Stroke. 2005;36(12):2742–2744. doi: 10.1161/01.STR.0000189748.52500.a7. [DOI] [PubMed] [Google Scholar]
  • 88.Arai K, Lo EH. Experimental models for analysis of oligodendrocyte pathophysiology in stroke. Exp Transl Stroke Med. 2009;1:6. doi: 10.1186/2040-7378-1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sasaki Y, Araki T, Milbrandt J. Stimulation of nicotinamide adenine dinucleotide biosynthetic pathways delays axonal degeneration after axotomy. J Neurosci. 2006;26(33):8484–8491. doi: 10.1523/JNEUROSCI.2320-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ho C, van der Veer E, Akawi O, Pickering JG. SIRT1 markedly extends replicative lifespan if the NAD+ salvage pathway is enhanced. FEBS Lett. 2009;583(18):3081–3085. doi: 10.1016/j.febslet.2009.08.031. [DOI] [PubMed] [Google Scholar]
  • 91.Li Y, Zhang Y, Dorweiler B, Cui D, Wang T, Woo CW, Brunkan CS, Wolberger C, Imai S, Tabas I. Extracellular Nampt promotes macrophage survival via a nonenzymatic interleukin-6/STAT3 signaling mechanism. J Biol Chem. 2008;283(50):34833–34843. doi: 10.1074/jbc.M805866200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, O’Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014;508(7494):55–60. doi: 10.1038/nature13165. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES